Language selection

Search

Patent 3111725 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 3111725
(54) English Title: 2,6-DIAMINO PYRIDINE COMPOUNDS
(54) French Title: COMPOSES DE 2,6-DIAMINO PYRIDINE
Status: Granted and Issued
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07D 40/14 (2006.01)
  • A61K 31/4427 (2006.01)
  • A61K 31/4439 (2006.01)
  • A61P 01/16 (2006.01)
  • A61P 03/10 (2006.01)
  • A61P 09/04 (2006.01)
  • C07D 40/04 (2006.01)
(72) Inventors :
  • DURHAM, TIMOTHY BARRETT (United States of America)
(73) Owners :
  • ELI LILLY AND COMPANY
(71) Applicants :
  • ELI LILLY AND COMPANY (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued: 2022-12-13
(86) PCT Filing Date: 2019-08-29
(87) Open to Public Inspection: 2020-03-12
Examination requested: 2021-03-03
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2019/048788
(87) International Publication Number: US2019048788
(85) National Entry: 2021-03-03

(30) Application Priority Data:
Application No. Country/Territory Date
62/726,520 (United States of America) 2018-09-04

Abstracts

English Abstract

The present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, and the use of compounds of Formula I for treating metabolic conditions, such as type 2 diabetes mellitus, heart failure, diabetic kidney disease, and non-alcoholic steatohepatitis.


French Abstract

La présente invention concerne un composé de formule I ou un sel pharmaceutiquement acceptable de celui-ci, et l'utilisation de composés de formule I pour le traitement d'états métaboliques, tels que le diabète sucré de type 2, l'insuffisance cardiaque, la néphropathie diabétique et la stéatohépatite non alcoolique.

Claims

Note: Claims are shown in the official language in which they were submitted.


26
CLAIMS
1. A compound of the formula:
H3
NC
I
F3C
Fonnula 1,
wherein n is 1 or 2;
or a pharmaceutically acceptable salt thereof.
2. The compound according to claim 1 wherein n is 1, or a pharmaceutically
acceptable salt thereof.
3. The compound according to claim 2 wherein the compound is:
CH3
NC
F3C
\._CO2H
or a pharmaceutically acceptable salt thereof.
4. The compound according to claim 1 wherein n is 2, or a pharmaceutically
acceptable salt thereof.
5. The compound according to claim 4 wherein the compound is:
Date Recue/Date Received 2021-04-28

27
C H 3
NC
F3C NO__/CO2 H
or a pharmaceutically acceptable salt thereof.
6. The compound according to claim 5 wherein the compound is:
CH 3
NC
F3CCO2 H
or a pharmaceutically acceptable salt thereof.
7. A use of a compound of any one of claims 1 to 6, or a phamiaceutically
acceptable salt thereof, for treating type 2 diabetes mellitus in a patient.
8. A use of a compound of any one of claims 1 to 6, or a phamiaceutically
acceptable salt thereof, in the manufacture of a medicament for the treatment
of diabetes
mellitus type 2.
9. A use of a compound of any one of claims 1 to 6, or a pharmaceutically
acceptable salt thereof, for treating heart failure in a patient.
10. A use of a compound according to any one of claims 1 to 6, or
a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament
for the
treatment of heart failure.
Date Recue/Date Received 2021-04-28

28
11. A use of a compound of any one of claims 1 to 6, or a pharmaceutically
acceptable salt thereof, for treating diabetic kidney disease in a patient.
12. A use of a compound of any one of claims 1 to 6, or a phamiaceutically
.. acceptable salt thereof, in the manufacture of a medicament for the
treatment of diabetic
kidney disease.
13. A use of a compound of any one of claims 1 to 6, or a pharmaceutically
acceptable salt thereof, for treating non-alcoholic steatohepatitis in a
patient.
14. A use of a compound of any one of claims 1 to 6, or a phamiaceutically
acceptable salt thereof, in the manufacture of a medicament for the treatment
of non-
alcoholic steatohepatitis.
15. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in therapy.
16. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in treating type 2 diabetes mellitus.
17. A compound, or a phafflaceutically acceptable salt thereof, according
to any
one of claims 1 to 6 for use in the manufacture of a medicament for the
treatment of type 2
diabetes mellitus.
18. A compound, or a phafflaceutically acceptable salt thereof, according
to any
one of claims 1 to 6 for use in treating heart failure.
Date Recue/Date Received 2021-04-28

29
19. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in the manufacture of a medicament for the
treatment of heart
failure.
20. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in treating diabetic kidney disease.
21. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in the manufacture of a medicament for the
treatment of diabetic
kidney disease.
22. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in treating non-alcoholic steatohepatitis.
23. A compound, or a pharmaceutically acceptable salt thereof, according to
any
one of claims 1 to 6 for use in the manufacture of a medicament for the
treatment of non-
alcoholic steatohepatitis.
24. A pharmaceutical composition comprising a compound, or a
pharmaceutically
acceptable salt thereof, according to any one of claims 1 to 6 with one or
more
pharmaceutically acceptable carriers, diluents, or excipients.
25. A process for preparing a pharmaceutical composition comprising
admixing a
compound, or a pharmaceutically acceptable salt thereof, according to any one
of claims 1 to
6 with one or more pharmaceutically acceptable carriers, diluents, or
excipients.
Date Recue/Date Received 2021-04-28

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-1-
2,6-DIAMINO PYRIDINE COMPOUNDS
The present invention relates to novel ketohexokinase (KHK) inhibitor
compounds, to
pharmaceutical compositions comprising the compounds and to the use of the
compounds for
.. the treatment of certain metabolic conditions, such as type 2 diabetes
mellitus (T2DM), heart
failure, diabetic kidney disease and non-alcoholic steatohepatitis (NASH).
Metabolic syndrome is commonly defined as a clustering of conditions that
reflect
over nutrition and a sedentary lifestyle and its manifestations include T2DM,
non-alcoholic
fatty liver disease (NAFLD), obesity, dyslipidemia, heart failure and kidney
disease.
T2DM is characterised by relative insulin deficiency caused by pancreatic 13-
cell
dysfunction and insulin resistance in target organs. It accounts for more than
90% of patients
with diabetes and leads to microvascular and macrovascular complications that
cause
profound psychological and physical distress to patients while placing a huge
burden on
health-care systems (Davies, M.J., et al.; Lancet, 389, 2239-2251, 2017).
Heart failure is a syndrome caused by structural or functional cardiac
abnormalities
that lead to elevated intracardiac pressures or a reduced cardiac output at
rest or during stress.
Heart failure is a leading and increasing cause of morbidity and mortality
worldwide
(Teerlink, J.R., et al.; Lancet, 390, 1981-1995, 2017).
Diabetic kidney disease develops in nearly half of patients with T2DM and is
the
leading cause of chronic kidney disease worldwide. Metabolic changes
associated with
diabetes leads to glomerular hyperfiltration, progressive albuminuria,
declining glomerular
filtration rate and ultimately end-stage renal disease (Alicic, R.Z., et at.;
Cl/n. I Am. Soc.
Nephrol., 12: 2032-2045, 2017).
NAFLD represents a spectrum of liver disease that can lead to progressive
NASH,
fibrosis, and ultimately hepatocellular carcinoma and liver failure. It is
estimated that in the
next 20 years, NAFLD will become the major cause of liver-related morbidity
and mortality
as well as the leading indication for liver transplantation (Bertot, L.C., et
at.; Int. I Mot. Sc.,
17(5), 774, 2016).

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-2-
KHK, also referred to as fructokinase, is the rate-limiting enzyme involved in
fructose
metabolism. It catalyses the phosphorylation of fructose to fructose-1-
phosphate (F1P),
causing concomitant depletion of cellular ATP levels. In contrast to glucose,
fructose
metabolism lacks feedback inhibition and it triggers accumulation of
downstream
intermediates involved in, for example, lipogenesis, gluconeogenesis and
oxidative
phosphorylation (Hannou, S.A., et al.; I Cl/n. Invest., 128(2), 544-555,
2018). This has
negative metabolic consequences which are associated with a number of serious
metabolic
disorders.
KHK exists in two alternatively spliced isoforms consisting of KHK-C and KHK-A
differing in exon 3. KHK-C is expressed primarily in the liver, kidney and
intestine, whereas
KHK-A is more ubiquitous. Mice deficient in both isoforms are fully protected
from
fructose-induced metabolic syndrome. However, the adverse metabolic effects
are
exacerbated in mice lacking KHK-A only (Ishimoto T, et al.; Proc. Natl. Acad.
Sci. USA,
109(11), 4320-4325, 2012).
Several epidemiologic and experimental studies have reported that increased
consumption of fructose, and more precisely increased fructose metabolism, may
play an
important role in the development of metabolic syndrome, in particular in the
development of
T2DM (Softic et al.; I Cl/n. Invest., 127(11), 4059-4074, 2017), heart failure
(Mirtschink,
P., et al.; Eur. Heart 1, 39, 2497-2505, 2018), diabetic kidney disease
(Cirillo, P., et al.;
Am. Soc. Nephrol., 20, 545-553, 2009) and NAFLD/NASH (Vos, M.B., et al.;
Hepatology,
57, 2525-2531, 2013). Targeting inhibition of KHK is expected to limit
fructose metabolism
and provide effective treatment options for a number of metabolic disorders.
US 2017/0183328 Al discloses substituted 3-azabicyclo[3.1.0]hexanes as KHK
inhibitors.
There is a need for alternate treatments for metabolic syndrome and associated
indications including T2DM, heart failure, diabetic kidney disease and NASH.
In particular,
there is a need for compounds having KHK inhibitory activity to provide
treatment options
for these diseases. There is a furthermore a need for potent KHK inhibitors
having properties

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-3-
which are important to the therapeutic use in humans, such as oral
bioavailability and a half-
life sufficient to support daily dosing, or a limited drug-drug interaction
profile.
Accordingly, the present invention provides a compound of Formula I:
O-CH3
NC
F3C
(V.PCO2H
Formula I
wherein n is 1 or 2;
or a pharmaceutically acceptable salt thereof.
Formula I includes all individual enantiomers and diastereomers thereof, as
well as
mixtures of enantiomers and racemates.
In a particular embodiment, a compound of the invention is a compound of the
formula:
C H3
NC
F3C
(VCO2H
or a pharmaceutically acceptable salt thereof.
In one embodiment, n is 1. In this embodiment, a compound of the invention is
a
compound of the formula:
H3
NC
F3C
OCO2H

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-4-
or a pharmaceutically acceptable salt thereof In a preferred embodiment, a
compound
of the invention is a compound of the formula:
CH3
NC
F3C
\CO2H
or a pharmaceutically acceptable salt thereof.
In another embodiment, n is 2. In this embodiment, a compound of the invention
is a
compound of the formula:
H3
NC
F3C, NajCO2H
or a pharmaceutically acceptable salt thereof. In a preferred embodiment, a
compound
of the invention is a compound of the formula:
CH3
NC
F3C, jCO2H
or a pharmaceutically acceptable salt thereof. In a further preferred
embodiment, a
compound of the invention is a compound of the formula:
CH3
NC
F3C,

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-5-
or a pharmaceutically acceptable salt thereof. In this embodiment, the
compound of
the invention is 2-[(3R)-1-[5-cyano-6-[(2S)-2-methylazetidin-1-y1]-4-
(trifluoromethyl)-2-
pyridyl]pyrrolidin-3-yl]acetic acid or a pharmaceutically acceptable salt
thereof.
In an embodiment, the present invention also provides a method of treating
T2DM in
a patient in need of such treatment comprising administering to the patient an
effective
amount of a compound of Formula I, or a pharmaceutically acceptable salt
thereof In a
preferred embodiment, the method comprises administering 2-[(3R)-1-[5-cyano-6-
[(2S)-2-
methylazetidin-1-y1]-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-3-yl]acetic
acid, or a
pharmaceutically acceptable salt thereof.
In an embodiment, the present invention also provides a method of treating
heart
failure in a patient in need of such treatment comprising administering to the
patient an
effective amount of a compound of Formula I, or a pharmaceutically acceptable
salt thereof.
In a preferred embodiment, the method comprises administering 2-[(3R)-1-[5-
cyano-6-[(2S)-
2-methylazetidin-1-y1]-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-3-yl]acetic
acid, or a
pharmaceutically acceptable salt thereof.
In an embodiment, the present invention also provides a method of treating
diabetic
kidney disease in a patient in need of such treatment comprising administering
to the patient
an effective amount of a compound of Formula I, or a pharmaceutically
acceptable salt
thereof In a preferred embodiment, the method comprises administering 2-[(3R)-
1-[5-
cyano-6-[(2S)-2-methylazetidin-1-y1]-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-
3-yl]acetic
acid, or a pharmaceutically acceptable salt thereof
In an embodiment, the present invention also provides a method of treating
NASH in
a patient in need of such treatment comprising administering to the patient an
effective
amount of a compound of Formula I, or a pharmaceutically acceptable salt
thereof In a
preferred embodiment, the method comprises administering 2-[(3R)-1-[5-cyano-6-
[(25)-2-
methylazetidin-1-y1]-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-3-yl]acetic
acid, or a
pharmaceutically acceptable salt thereof.
In an embodiment, the present invention also provides a method of treating
chronic
kidney disease in a patient in need of such treatment comprising administering
to the patient

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-6-
an effective amount of compound of Formula I, or a pharmaceutically acceptable
salt thereof.
In a preferred embodiment, the method comprises administering 2-[(3R)-1-[5-
cyano-6-[(2S)-
2-methylazetidin-l-y1]-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-3-yl]acetic
acid, or a
pharmaceutically acceptable salt thereof.
In an embodiment, the present invention further provides a method of treating
a
disease selected from the group consisting of metabolic syndrome, NAFLD,
obesity, diabetic
complications for example diabetic retinopathy, cardiovascular disease,
coronary artery
disease, and dyslipidemia, in a patient in need of such treatment, comprising
administering to
the patient an effective amount of a compound of Formula I, or a
pharmaceutically
acceptable salt thereof. In a preferred embodiment, the method comprises
administering 2-
[(3R)-1-[5-cyano-6-[(2S)-2-methylazetidin-1-y1]-4-(trifluoromethyl)-2-
pyridyl]pyrrolidin-3-
yl]acetic acid, or a pharmaceutically acceptable salt thereof.
Furthermore, in an embodiment, this invention provides a compound of Formula
I, or a
pharmaceutically acceptable salt thereof, for use in therapy. In a particular
embodiment, the
present invention provides a compound of Formula I, or a pharmaceutically
acceptable salt
thereof, for use in treating T2DM. In a particular embodiment, the present
invention
provides a compound of Formula I, or a pharmaceutically acceptable salt
thereof, for use in
treating heart failure. In a particular embodiment, the present invention
provides a compound
of Formula I, or a pharmaceutically acceptable salt thereof, for use in
treating diabetic kidney
disease. In a particular embodiment, the present invention provides a compound
of Formula
I, or a pharmaceutically acceptable salt thereof, for use in treating NASH. In
a particular
embodiment, the present invention also provides a compound of Formula I, or a
pharmaceutically acceptable salt thereof, for use in treating chronic kidney
disease. In an
embodiment, the invention also provides a compound of Formula I, or a
pharmaceutically
acceptable salt thereof, for use in treating metabolic syndrome, NAFLD,
obesity, diabetic
complications for example diabetic retinopathy, cardiovascular disease,
coronary artery
disease, or dyslipidemia. In a preferred embodiment, the compound of Formula
Tin the
therapeutic uses above is 2-[(3R)-145-cyano-6-[(25)-2-methylazetidin-1-y1]-4-

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-7-
(trifluoromethyl)-2-pyridyl]pyrrolidin-3-yl]acetic acid, or a pharmaceutically
acceptable salt
thereof
Furthermore, in an embodiment, this invention provides the use of a compound
of
Formula I, or a pharmaceutically acceptable salt thereof, for the manufacture
of a
medicament for treating T2DM. In an embodiment, the present invention provides
the use of
a compound of Formula I, or a pharmaceutically acceptable salt thereof, for
the manufacture
of a medicament for treating heart failure. In an embodiment, the present
invention provides
the use of a compound of Formula I, or a pharmaceutically acceptable salt
thereof, for the
manufacture of a medicament for treating diabetic kidney disease. In an
embodiment, the
present invention provides the use of a compound of Formula I, or a
pharmaceutically
acceptable salt thereof, for the manufacture of a medicament for treating
NASH. In an
embodiment, the present invention provides the use of a compound of Formula I,
or a
pharmaceutically acceptable salt thereof, for the manufacture of a medicament
for treating
chronic kidney disease. In an embodiment, the invention also provides the use
of a
compound of Formula I, or a pharmaceutically acceptable salt thereof, for the
manufacture of
a medicament for treating metabolic syndrome, NAFLD, obesity, diabetic
complications for
example diabetic retinopathy, cardiovascular disease, coronary artery disease,
or
dyslipidemia. In a preferred embodiment, the compound of Formula I is 2-[(3R)-
1-[5-cyano-
6-[(2S)-2-methylazetidin-1-y1]-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-3-
yl]acetic acid, or a
pharmaceutically acceptable salt thereof.
In an embodiment, the invention further provides a pharmaceutical composition
comprising a compound of Formula I, or a pharmaceutically acceptable salt
thereof, with one
or more pharmaceutically acceptable carriers, diluents, or excipients. In an
embodiment, the
invention further provides a process for preparing a pharmaceutical
composition comprising
admixing a compound of Formula I, or a pharmaceutically acceptable salt
thereof, with one
or more pharmaceutically acceptable carriers, diluents, or excipients.
As used herein, the terms "treating" or "to treat" includes restraining,
slowing,
stopping, or reversing the progression or severity of an existing symptom or
disorder.

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-8-
As used herein, the term "patient" refers to a mammal. Preferably, the patient
is
human.
As used herein, the term "effective amount" refers to the amount or dose of a
compound of Formula I, or a pharmaceutically acceptable salt thereof, which,
upon single or
multiple dose administration to the patient, provides the desired effect in
the patient under
diagnosis or treatment.
An effective amount can be determined by one skilled in the art by the use of
known
techniques and by observing results obtained under analogous circumstances. In
determining
the effective amount for a patient, a number of factors are considered,
including, but not
limited to: the species of patient; its size, age, and general health; the
specific disease or
disorder involved; the degree of or involvement or the severity of the disease
or disorder; the
response of the individual patient; the particular compound administered; the
mode of
administration; the bioavailability characteristics of the preparation
administered; the dose
regimen selected; the use of concomitant medication; and other relevant
circumstances. The
compounds of the present invention are effective at a dosage per day that
falls within the
range of about 0.1 to about 15 mg/kg of body weight.
The compounds of the present invention are formulated as pharmaceutical
compositions administered by any route which makes the compound bioavailable.
Preferably, such compositions are for oral administration. Such pharmaceutical
compositions
and processes for preparing same are well known in the art (See, e.g.,
Remington, J. P.,
"Remington: The Science and Practice of Pharmacy", L.V. Allen, Editor, 22nd
Edition,
Pharmaceutical Press, 2012).
The compounds of Formula I and the pharmaceutically acceptable salts thereof
are
particularly useful in the treatment methods of the invention, with certain
configurations
being preferred. The following list of compounds of the present invention
describe such
configurations. It will be understood that these preferences are applicable to
the compounds
of the invention, as well as the treatment methods, therapeutic uses and
pharmaceutical
compositions.
Compounds of the present invention include:

CA 03111725 2021-03-03
WO 2020/051058
PCT/US2019/048788
-9-
C H3
NC
F3C
(VCO2H
Formula Ia
C H3
NC
F3C
(VCO2H
Formula lb
and pharmaceutically acceptable salts thereof
Further compounds of the present invention include:
CH3
NC
F3C)LN
\CO2H
Formula Ha
NC
F3C
\._3CO2H
Formula Ilb
and pharmaceutically acceptable salts thereof
Further compounds of the present invention include:

CA 03111725 2021-03-03
WO 2020/051058
PCT/US2019/048788
-10-
CH3
NCLN
F3C CO2H
Formula Ma'
CH3
NC
Formula Ma"
H3
NC
C H02
Formula IIIb'
O---C H3
NC
F3CNI.D/C 2
Formula Mb"
and pharmaceutically acceptable salts thereof
Although the present invention contemplates all individual enantiomers and
diasteromers, as well as mixtures thereof, including racemates, compounds of
Formula Ia,
Formula Ha and Ma", and pharmaceutically acceptable salts thereof, are
particularly
preferred.

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-11-
Individual enantiomers may be separated or resolved by one of ordinary skill
in the
art at any convenient point in the synthesis of compounds of the invention, by
methods such
as selective crystallization techniques, chiral chromatography (See for
example, J. Jacques, et
at., "Enantiomers, Racemates, and Resolutions", John Wiley and Sons, Inc.,
1981, and E.L.
Eliel and S.H. Wilen," Stereochemistry of Organic Compounds", Wiley-
Interscience, 1994),
or supercritical fluid chromatography (SFC) (See for example, T. A. Berger;
"Supercritical
Fluid Chromatography Primer," Agilent Technologies, July 2015).
A pharmaceutically acceptable salt of the compounds of the invention can be
formed,
for example, by reaction of an appropriate neutral form of a compound of the
invention and
an appropriate pharmaceutically acceptable acid or base in a suitable solvent
under standard
conditions well known in the art (See, for example, Bastin, R.J., et at.; Org.
Process. Res.
Dev., 4, 427-435, 2000 and Berge, S.M., et al.; I Pharm. Sci., 66, 1-19,
1977).
The compounds of the present invention, or salts thereof, may be prepared by a
variety of procedures known to one of ordinary skill in the art, some of which
are illustrated
in the schemes, preparations, and examples below. The products of each step in
the schemes
below can be recovered by conventional methods well known in the art,
including extraction,
evaporation, precipitation, chromatography, filtration, trituration, and
crystallization. In the
schemes below, all substituents unless otherwise indicated, are as previously
defined. The
reagents and starting materials are readily available to one of ordinary skill
in the art.
Without limiting the scope of the invention, the following schemes,
preparations, and
examples are provided to further illustrate the invention. In addition, one of
ordinary skill in
the art appreciates that compounds of Formula I may be prepared by using
starting material
or intermediate with the corresponding desired stereochemical configuration
which can be
prepared by one of skill in the art.
Certain abbreviations are defined as follows: "ABT" refers to 1-
aminobenzotriazole;
"ACN" refers to acetonitrile; "BSA" refers to bovine serum albumin; "CAS#"
refers to
Chemical Abstracts Registry number; "DCM" refers to methylene chloride or
dichloromethane; "DIPEA" refers to diisopropylethylamine; "DMEM" refers to
Dulbecco's
Modified Eagle's medium; "DMSO" refers to dimethyl sulfoxide; "ELSD" refers to

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-12-
Evaporative light scattering detector; "ES/MS" refers to Electrospray Mass
Spectrometry;
"Et0Ac" refers to ethyl acetate; "Et0H" refers to ethanol or ethyl alcohol;
"FBS" refers to
fetal bovine serum; "h" refers to hour or hours; "HPLC" refers to high-
performance liquid
chromatography; "Me" refers to methyl; "Me0H" refers to methanol; "MTBE"
refers to
methyl-tert-butyl ether; "min" refers to minute or minutes; "m/z" refers to
mass-to-charge
ratio; "PBS" refers to phosphate buffered saline; "Ph" refers to phenyl; "RBF"
refers to
round bottom flask; "RT" refers to room temperature; "SCX" refers to selective
cation
exchange; "SFC" refers to Supercritical Fluid Chromatography; "THF" refers to
tetrahydrofuran.
Scheme 1
CI CI
NCLN NC
HiN1
Step 1 LN
C
+ O2C H3
F3C F3c-
co2c H3
1 2 3 4
CH3 C H3
Step 2 NC Step 3
NC
I
F3C
( CO2CH3 F3C-
( CO2H
n n
5 Ia
Scheme 1 depicts the general preparation of the compounds of Formula I. In
Step 1,
3-cyano-2,6-dichloro-4-(trifluoromethyl)pyridine (1) and a cyclic amine (2)
are reacted in the
presence of a base, such as NaHCO3 or DIPEA, in Et0H or Me0H to yield an
aminopyridine
(3). Alternatively, the reaction solvent for this step can be DCM. In Step 2,
compound 3 is
reacted with 2-methyl azetidine (4) at elevated temperature and in the
presence of a base,
such as NaHCO3 or DIPEA, in Et0H or Me0H to give a diaminopyridine (5).
Alternatively,

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-13-
the reaction solvent for this step can be THF. In Step 3, the ester moiety is
hydrolysed using a
base such as NaOH or LiOH in Me0H or THF at elevated temperature to give a
compound
of Formula Ia. Alternatively, this step may be performed using the same
reagents in a
microwave reactor.
Preparations and Examples
The following Preparations and Examples further illustrate the invention and
represent typical synthesis of the compound of the invention. The reagents and
starting
materials are readily available or may be readily synthesized by one of
ordinary skill in the
art. It should be understood that the Preparations and Examples are set forth
by way of
illustration and not limitation, and that various modifications may be made by
one of
ordinary skill in the art.
LC-ES/MS is performed on an AGILENT HP1100 liquid chromatography system.
Electrospray mass spectrometry measurements (acquired in positive and/or
negative mode)
are performed on a Mass Selective Detector quadrupole mass spectrometer
interfaced to an
HPLC which may or may not have an ELSD. LC-MS conditions (low pH): column:
PHENOMENEX GEMINI NX C18 2.0 x 50 mm 3.0 m, 110 A; gradient: 5-95% B in
1.5 min, then 95% B for 0.5 min column temperature: 50 C +/-10 C; flow rate:
1.2
mL/min; 1 IAL injection volume; Solvent A: deionized water with 0.1% HCOOH;
Solvent B:
ACN with 0.1% formic acid; wavelength 200-400 nm and 212-216 nm. If the HPLC
is
equipped with an ELSD the settings are 45 C evaporator temperature, 40 C
nebulizer
temperature, and 1.6 SLM gas flow rate. Alternate LC-MS conditions (high
pH): column: Waters xBridge C18 column 2.1x50 mm, 3.5 m; gradient: 5-95% B
in 1.5
min, then 95% B for 0.50 min; column temperature: 50 C +/-10 C; flow rate:
1.2 mL/min;
1pL injection volume; Solvent A: 10 mM NH4HCO3 pH 9; Solvent B: ACN ;
wavelength:
200-400 nm and 212-216nm; if had ELSD: 45 C evaporator temp, 40 C nebulizer
temp, and
1.60 SLM gas flow rate.

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-14-
Preparation 1
(2S)-1-Benzhydry1-2-methyl-azetidine [(1R,4S)-7,7-dimethy1-2-oxo-norbornan-1-
yl]methanesulfonic acid salt
CH3
H3C473H
Ph,(
CH3 --O
Ph
Assemble a 2000 mL 3-neck RBF with an addition funnel, nitrogen inlet and a
thermometer adapter. Purge the vessel with nitrogen and add (3R)-butane-1,3-
diol (25 g,
277.4 mmol), DIPEA (127 ml, 731 mmol) and ACN (556 m1). Cool to -30 C. Add
trifluoromethanesulfonic anhydride (101 mL, 601 mmol) dropwise over 3 h such
that the
internal temperature is maintained between -35 and -30 C. After the
completion of the
addition, stir for 10 min at -35 to -30 C. Add trifluoromethanesulfonic
anhydride (1.9 mL,
11 mmol) dropwise over 5 min such that the internal temperature is maintained
between -35
and -30 C. After the completion of the addition, stir for 10 min at -35 to -
30 C. Add
DIPEA (127 mL, 731 mmol) dropwise over 15 min such that the internal
temperature is
maintained between -35 and -30 C. After the completion of the addition, stir
for 10 min at -
35 to -30 C. In a separate flask under nitrogen, dissolve
aminodiphenylmethane (48.0 mL,
270 mmol) in ACN (49 mL, 935 mmol) and transfer the resulting solution to the
addition
funnel. Add the amine solution to the cold triflate dropwise over 40 min such
that the internal
temperature is maintained between -20 to -35 C. After the completion of the
addition, stir
for 30 min at -35 to -30 C. Transfer the reaction to a water bath and allow
it to slowly warm
over 30 min. Remove the bath and allow the reaction to warm to RT over 30 min.
Transfer
the vessel to a heating mantle and warm the reaction to 45 C for 30 min, then
cool to RT.
Pour the resulting mixture into 1200 mL of water and extract with toluene (400
mL x 3).
Combine the extracts, wash with water, sat. aq. NaCl solution, dry over
anhydrous Na2SO4,
filter and concentrate on a rotary evaporator. Dry the material under vacuum
overnight.
Dissolve the residue in DCM (400 mL). Prepare a silica pad on a fritted funnel
and
equilibrate it with 1:1 heptane/Et0Ac. Load the product solution onto the
silica pad and wash
with 1600 mL of 1:1 heptane/Et0Ac. Concentrate the filtrate to give a red oil.
Dissolve the

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-15-
oil in Me0H (250 mL) and place the flask in a water bath (-10 C). Add L(-)-
camphorsulfonic acid (61.6 g, 265 mmol) portion-wise keeping the internal
temperature
below 20 C. Stir the resulting mixture for 15 min and then concentrate on a
rotary evaporator
to give a brown foam. Dry the foam on a vacuum pump for 2 h. Dissolve the foam
in 130
mL of DCM. Attach an addition funnel to the flask. Use the funnel to slowly
add 1100 mL of
Et0Ac to the stirring solution. Transfer the resulting mixture to a 4000 mL
beaker and stir
open to the atmosphere overnight. Cool the beaker in an ice bath for 10 min.
Collect the
precipitate in a fritted funnel by vacuum filtration washing with a minimal
amount of ice-
cold Et0Ac. Dry the solid on the frit for 2 h. Dissolve the resulting white
solid in a minimal
amount of DCM, transfer to a 2000 mL beaker and then dilute slowly with Et0Ac
until the
clear solution starts to become cloudy. Stir the suspension for 4 h while open
to the
atmosphere. Collect the solids by vacuum filtration using a fritted funnel and
dry on the frit
overnight to give the title compound (111.8 g, 238.06 mmol, 86% Yield) as a
white solid. 1-H
NMR (400 MHz, d6-DMS0): 10.54-10.47 (m, 1H), 7.61 (d, J= 7.3 Hz, 5H), 7.47-
7.37 (m,
7H), 5.85 (d, J= 10.3 Hz, 1H), 4.68-4.61 (m, 1H), 3.91-3.83 (m, 2H), 3.37 (s,
8H), 2.99 (d, J=
14.6 Hz, 1H), 2.77-2.68 (m, 1H), 2.51-2.44 (m, 4H), 2.30-2.16 (m, 2H), 1.91-
1.81 (m, 2H),
1.42-1.28 (m, 3H), 1.08 (s, 3H), 1.01 (d, J= 6.6 Hz, 3H), 0.77 (s, 4H); >98%
ee [HPLC:
Chiralcel OJ (10 cm x 4.6 mm, 5 m), 5 mL/min, 40 C isocratic 10% Et0H (0.2%
PrNH2)/CO21.
Preparation 2
[(1R,4S)-7,7-Dimethy1-2-oxo-norbornan-1-yl]methanesulfonate (2S)-2-
methylazetidin-1-ium
salt
01-13
H3Cv3H
0
H C H3
To a 2250 mL Parr vessel add 20 wt% Pd(OH)2 on carbon (6.62 g). Purge the
bottle
with nitrogen and add 250 mL of Me0H. To the resulting suspension, slowly add
(2S)-1-
b enzhydryl -2-methyl -azeti dine [(1R,4 S)-7,7-dimethy1-2-oxo-norb ornan-1-

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-16-
yl]methanesulfonic acid salt (111 g, 236 mmol) dissolved in 250 mL of Me0H.
Seal the
vessel. Purge with nitrogen followed by hydrogen and pressurize to 60 PSI.
Vigorously shake
the reaction vessel in a Parr Shaker apparatus for 15 h at RT. Purge the
vessel with nitrogen
and then filter the reaction mixture through a pad of celite, washing with
Me0H. Concentrate
the filtrate to give a white solid and dry under vacuum. Suspend the solid in
780 mL of 1:1
MTBE/Et0Ac and heat the mixture to 65 C for 20 h then cool to RT and stir
overnight.
Collect the solids by filtration. Suspend the solids in 380 mL of MTBE and
stir at RT for 24
h. Collect the white solid by filtration to give the title compound (41.5 g,
136.78 mmol, 58%
Yield). 1H NMR (400 MHz, d6-DMS0): 8.68-8.55 (m, 1H), 4.51-4.42 (m, 1H), 3.91-
3.75
(m, 1H), 3.36 (s, 3H), 2.91 (d, J= 14.6 Hz, 1H), 2.69-2.61 (m, 1H), 2.52-2.46
(m, 2H), 2.28-
2.22 (m, 1H), 2.17-2.10 (m, 1H), 1.96 (t, J= 4.5 Hz, 1H), 1.89-1.79 (m, 1H),
1.43 (d, J= 6.7
Hz, 2H), 1.36-1.26 (m, 1H), 1.05 (s, 2H), 0.75 (s, 2H).
Preparation 3
Methyl 2-[(3R)-1-[6-chloro-5-cyano-4-(trifluoromethyl)-2-pyridyl]pyrrolidin-3-
yl]acetate
CI
NC
No......"CO2CH3
F3C-
To a RBF add 3-cyano-2,6-dichloro-4-(trifluoromethyl)pyridine (123 mmol, 29.6
g)
and Et0H (230 mL). Cool the mixture to 0 C. Add NaHCO3 (368 mmol, 31 g)
followed by a
solution of methyl (R)-pyrrolidine-3-acetate hydrochloride (23 g, 123 mmol) in
Et0H (230
mL). Allow the resulting mixture to warm to RT overnight. Evaporate the
reaction mixture to
dryness on a rotary evaporator. Add water (200 mL) and extract with MTBE (2 x
200 mL).
Combine the extracts and evaporate to dryness. Purify by chromatography on
silica gel using
hexane/MTBE (gradient from 20 to 70%) to give the title compound (34.7 g,
99.89 mmol,
81% Yield) as a white solid. ES/MS (m/z): 348.0, 350.0 [M+H]t

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-17-
Preparation 4
Methyl 2-[(3R)-1-[5-cyano-6-[(2S)-2-methylazetidin-l-y1]-4-(trifluoromethyl)-2-
pyridyl]pyrrolidin-3-yl]acetate
(>.'C H3
NC
F3C Nc.}..."CO2C H3
'
To a RBF add methyl 2-[(3R)-1-[6-chloro-5-cyano-4-(trifluoromethyl)-2-
pyridyl]pyrrolidin-3-yl]acetate (25.5 g, 73.3 mmol), [(1R,4S)-7,7-dimethy1-2-
oxo-norbornan-
1-yl]methanesulfonate (2S)-2-methylazetidin-1-ium salt (88.0 mmol, 26.7 g),
Me0H (255
mL), and NaHCO3 (220 mmol, 18.5 g). Stir the mixture at 65 C for 16 h. Add
[(1R,4S)-7,7-
dimethy1-2-oxo-norbornan-l-yl]methanesulfonate (2S)-2-methylazetidin-1-ium
salt (22.0
mmol, 6.68 g) and NaHCO3 (147 mmol, 12.3 g) and continue stirring for 32 h.
Remove the
solvent on a rotary evaporator. To the residue add water (300 mL) and extract
with MTBE (2
x 200 mL). Combine the extracts and dry over anhydrous MgSO4, filter through
silica gel,
and concentrate to dryness to give the title compound (28.0 g, 73.2 mmol, 99%
Yield) as a
white solid. ES/MS (m/z): 383.2 [M+H]t
Preparation 5
Methyl 2-[1-[6-chloro-5-cyano-4-(trifluoromethyl)-2-pyridyl]azetidin-3-
yl]acetate
CI
NC
F3C-
CO2 C H3
To a solution of 3-cyano-2,6-dichloro-4-(trifluoromethyl)pyridine (500 mg,
2.03
mmol) in Et0H (15 mL) add NaHCO3 (0.549 g, 6.51 mmol) and methyl 2-(azetidin-3-
yl)acetate trifluoroacetic acid salt (0.494 g, 2.03 mmol). Stir the mixture at
RT for 16 h.
Dilute the reaction mixture with sat. aq. NaCl solution (30 mL). Extract with
Et0Ac (20 mL

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-18-
x 3). Combine the extracts and wash with sat. aq. NaCl solution (30 mL), dry
over anhydrous
Na2SO4, filter, and concentrate. Purify the residue by column chromatography
on silica gel
eluting with Et0Ac in petroleum ether (gradient 0-30%) to give the title
compound (470 mg,
1.41 mmol, 66% Yield) as a white solid. ES/MS (m/z) = 333.9 [M+H]t
Preparation 6
Methyl 2-[1-[5-cyano-6-[(2S)-2-methylazetidin-1-y1]-4-(trifluoromethyl)-2-
pyridyl]azetidin-
3-yl]acetate
NC
F3C
CO2 C H3
To a solution of methyl 2-[1-[6-chloro-5-cyano-4-(trifluoromethyl)-2-
pyridyl]azetidin-3-yl]acetate (200 mg, 0.569 mmol) in Et0H (6 mL) add NaHCO3
(0.173 g,
2.05 mmol) and [(1R,45)-7,7-dimethy1-2-oxo-norbornan-1-yl]methanesulfonate
(25)-2-
methylazetidin-1-ium salt (0.194 g, 0.626 mmol). Heat the mixture to 80 C for
16 h. Dilute
with water (50 mL) and extract with Et0Ac (40 mL x 3). Combine the extracts
and wash
with sat. aq. NaCl solution (50 mL), dry over anhydrous Na2SO4 filter and
concentrate.
Purify the residue by column chromatography on silica gel (0%-10%, Et0Ac in
petroleum
ether) to give the title compound (176 mg, 0.46 mmol, 77% Yield) as a white
solid. ES/MS
(m/z): 383.1 [M+H]t
Example 1
2-[(3 R)-1-[5-cyano-6-[(2S)-2-methylazetidin-l-y1]-4-(trifluoromethyl)-2-
pyridyl]pyrrolidin-
3-yl]acetic acid

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-19-
CH3
NC
CO2H
F3 C-
To a RBF add methyl 2-[(3R)-145-cyano-6-[(2S)-2-methylazetidin-1-y1]-4-
(trifluoromethyl)-2-pyridyl]pyrrolidin-3-yl]acetate (26.5 g, 69.3 mmol), Me0H
(265 mL),
and 2 M aq. NaOH solution (416 mmol, 208 mL). Stir the mixture at 60 C for 3
h. Cool to
RT and remove Me0H on a rotary evaporator. Acidify the aqueous phase to pH 3-4
using
concentrated aq. HC1. Extract with Et0Ac (400 mL). Wash the organic layer with
water (200
mL), dry over anhydrous MgSO4, filter, and concentrate to dryness to give the
title
compound (22.5 g, 61.11 mmol, 88% Yield) as a white foam. ES/MS (m/z): 369.2
[M+H]t
Example 2
2-[1-[5-cyano-6-[(25)-2-methylazetidin-1-y1]-4-(trifluoromethyl)-2-
pyridyl]azetidin-3-
yl]acetic acid
C H3
NCLN
F3C-
CO2H
To a mixture of methyl 2-[1-[5-cyano-6-[(25)-2-methylazetidin-1-y1]-4-
(trifluoromethyl)-2-pyridyl]azetidin-3-yl]acetate (176 mg, 0.478 mmol) in THF
(5.00 mL)
and water (1.00 mL) add LiOH (0.04 g, 0.955 mmol). Stir the resulting mixture
at RT for 2
h. Dilute with Et0Ac (4 mL) and extract the aqueous layer with Et0Ac (2 mL x
3).
Combine the organic extracts and wash with sat. aq. NaCl solution (3 mL x 2),
dry over
anhydrous Na2SO4, filter, concentrate to afford the title compound (128 mg,
0.36 mmol, 78%
Yield) as a white solid. ES/MS (m/z) 354.9 [M+H]+.

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-20-
Assays
KHK Enzyme Activity Assay for Human KHK-C and Human KHK-A
The intrinsic potency of the compounds may be measured using an enzymatic
assay
which measures the production of FlP. Compounds are prepared in DMSO and
tested in a
10-point concentration curve, to create 3-fold serial dilutions of the
compounds in a 96-well
plate ranging from 20 i.tM to 1.02 nM. Enzyme is prepared in assay buffer [50
mM 4-(2-
hydroxyethyl)piperazine-1-ethanesulfonic acid (HEPES), 10 mM potassium
chloride, 100
mM magnesium chloride, 2 mM tris(2-carboxyethyl)phosphine (TCEP), 0.01% n-
octyl
glucoside] and incubated with compounds at RT for 15 min. The reaction is
carried out in
100 [IL volumes containing substrate concentrations of fructose (250 [LM for
KHK-C assay
and 1.25mM for KHK-A assay) and ATP (150 [LM for both isoforms); which are
further
incubated at RT for 20 min. The reaction is then halted by the addition of
stop buffer;
consisting of 0.2% formic acid and 11.tg/m113C6-fructose-6-phosphate (1-3C6-
F6P) internal
standard. Plates are stored in -20 C until RapidFire MS analysis.
RapidFire MS Analysis for Quantitation of FlP:
An Agilent 300 RapidFire automated extraction system (Agilent, Santa Clara,
CA)
with three HPLC quaternary pumps is coupled to an Agilent 6495 triple
quadrupole mass
spectrometer (Agilent Technologies, Santa Clara, CA) equipped with an
electrospray
ionization (ESI) interface source. The RapidFire Mass Spec system is equipped
with a
reusable RapidFire C18 (type C) solid-phase extraction (SPE) cartridge
(G9205A).
Solvent A, used for sample loading and washing, is 6 mM octylamine (Acros
Organics 129495000) brought to pH 5.0 using acetic acid. Solvent B, used for
sample
elution, is 20% water in ACN containing 0.1% formic acid. Samples are
sequentially
analyzed by aspirating 10 [IL onto the collection loop under vacuum directly
from multiwell
.. plates. The 10 [IL of sample is loaded onto the C18 cartridge and washed
using solvent A at a
flow rate of 1.25 mL/min for 5000 ms. The retained analytes are then eluted to
the mass
spectrometer using solvent B at a flow rate of 1.25 mL/min for 5000 ms. The
system is re-
equilibrated using solvent A at a flow rate of 1.25 mL/min for 2000 ms.

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-21-
The triple quadrupole mass spectrometer is equipped with an ESI source and
analytes
are monitored using selected reaction monitoring (SRM) in negative mode EM-H]-
. F1P is
monitored at m/z 259.02/96.9 and 13C6-fructose-6-phospate is monitored at m/z
264.99/97.
The area ratio values for FlP is calculated using 13C6-fructose-6-phospate as
internal
standard.
The compounds of Examples 1 and 2 were tested essentially as described above:
Table 1
Example Number hKHK-C ICso (nM) hKHK-A ICso (nM)
1 9 19
2 27 37
These results demonstrate that the compounds of Examples 1 and 2 inhibit the
enzymatic activity of both KHK-C and KHK-A.
KHK Cellular Activity Assay
Potency can be measured using a cellular assay for the inhibition of the
conversion of
Fructose to FlP by cellular KHK. HepG2 cells are plated on 96-well cell
culture plates in
growth media [Dulbecco's Modified Eagle's medium (DMEM) high glucose, 10% heat-
inactivated fetal bovine serum (HI FBS), lx Penicillin/streptomycin] and
allowed to attach
overnight in a 37 C incubator. The growth media is washed and replaced with
assay media
consisting of Gibco OptiMEM 1 Reduced Serum Medium, 0.1% Casein, 8.33 mM D-
Fructose-13C6, and compound concentrations ranging from 10011M to 0.005111M
(10-point
concentration curve). Plates are incubated at 37 C for 3 h, after which assay
media is
aspirated from the cell wells. Stop solution consisting of 80% methanol, 2 mM
ammonium
acetate, and 50 ng/mL fructose-6-phosphate-13C6 is then added to the cells.
Plates are stored
in -20 C until RapidFire MS analysis (described above).
The compounds of Examples 1 and 2 were tested essentially as described above:

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-22-
Table 2
Example Number HepG2 IC50
1 127
2 316
These results demonstrate that the compounds of Examples 1 and 2 inhibit the
metabolism of fructose to FlP.
Liquid Chromatograph with Tandem Mass Spectrometry (LC-MS/MS) Method for
Pharmacokinetic Assays: Samples are extracted using a protein precipitation by
adding
1804, of MeOH:ACN (1:1, v/v) containing an internal standard and 25 !IL
(rodent samples)
or 50 !IL (non-rodent samples) of plasma. Samples are then diluted with
MeOH:Water (1:1,
v/v) to get concentrations within standard curve range. Diluted samples are
analyzed by LC-
MS/MS using a Sciex API 4000 triple quadrupole mass spectrometer (Applied
Biosystems/MDS; Foster City, CA) equipped with a TurboIonSpray interface, and
operated
in positive ion mode. The analytes are chromatographically separated using a
Thermo
Betasil C18 Sum 20X2.1mm Javelin column (rodent samples) or Advantage ECHIELON
C18 4um 20 mm x 2.1 mm ID column (non-rodent samples). LC conditions are
Water/1 M
ammonium bicarbonate, (2000:10, v/v) (Mobile Phase A), and Me0H/1 M ammonium
bicarbonate, (2000:10, v/v) (Mobile Phase B).
Pharmacokinetics in Mice
The in vivo pharmacokinetic properties and involvement of OATP1A/1B
transporter
in disposition of Example 1 are demonstrated using FVB wild type mice and
OATP1A/1B
knockout mice (Taconic #10707) (fasted; n=4/genotype). Example 1 is
administered by a
.. single intravenous (IV; lmg/kg; volume of lmL/kg) dose in vehicle. Blood is
collected from
each animal at 0, 0.08, 0.25, 0.5, 1, 2, 4, 8, 12, and 24 h post-dosage.
Liver, spleen, and

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-23-
pancreas is collected, weighed and perfused at 24 h post-dosage. The plasma
and tissue
concentrations of Example 1 are determined by a LC-MS/MS method as described
above.
In FVB mice, Example 1 has 5.43 hour half-life, a mean clearance of 6.91
mL/hr/kg,
a volume of distribution of 2.74 L/kg, with a mean liver unbound partition
coefficient (K)
of 67.5. In OATP1A/1B knockout mice, Example 1 has 9.36 hour half-life, a mean
clearance
of 1.34 mL/hr/kg, a volume of distribution of 0.986 L/kg, with a mean liver
unbound
partition coefficient (K) of 24.2. This data shows OATP is involved in the
hepatic uptake
of Example 1 in the mouse and that involvement of this transporter affects
clearance.
Pharmacokinetics in Dogs
The in vivo pharmacokinetic properties of Example 1 are demonstrated using
Beagle
Dogs (fed, n=3-4). Example 1 is administered by a single oral (PO; 3 mg/kg;
volume of 2
mL/kg) or intravenous (IV; 1 mg/kg; volume of 1 mL/kg) dose in vehicle. Blood
is collected
from each animal at 0, 0.03 (IV group only), 0.08 (IV), 0.25, 0.5, 1, 2, 4, 8,
12, 24, 32 (IV),
48 (IV), and 72 (IV) h post-dosage. The plasma concentrations of Example 1 are
determined
by a LC-MS/MS method as described above.
For PO doses, Example 1 mean half-life is 6.9 hours and bioavailability is
¨93%. For
IV doses, Example 1 mean half-life is 7.4 hours and the mean clearance is 2.52
mL/hr/kg
with a low volume of distribution (1.33 L/kg). This data shows Example 1 has
low total
clearance, low volume of distribution and high oral bioavailability in dogs.
The characterization of major elimination pathways in vivo are demonstrated
using
bile duct cannulated (BDC) Beagle Dogs (fed; n=3). Example 1 is administered
by a single
intravenous (IV; 1 mg/kg; volume of 1 mL/kg) dose in vehicle. Blood is
collected from each
animal at 0, 0.033, 0.083, 0.25, 0.5, 1, 2, 4, 8, 12, 24, 32, 48 and 72 h post-
dosage. Bile is
collected from each animal at 1, 2, 3, 4, 5, 6, 12, 18, 24, 32, 48 and 72 h
post-dosage. Urine
is collected at 12, 24, 48, and 72 h and feces collected at 24, 48, and 72 h
post-dosage. The
plasma, bile, urine and feces concentrations of Example 1 are determined by a
LC-MS/MS
method as described above.

CA 03111725 2021-03-03
WO 2020/051058
PCT/US2019/048788
-24-
Example 1 mean half-life is 2.9 hours and the mean clearance is 4.69 mL/hr/kg
with a
low volume of distribution (0.546 L/kg). Example 1 urine levels are negligible
and ¨10% of
the administered IV dose is recovered in bile. This data shows Example 1 has
low renal and
biliary elimination. Overall, the elimination half-life in bile cannulated
dogs, 2.9 hr, is faster
than the half-life measured in intact dogs, 7.4 hr, suggesting enterohepatic
recirculation.
Pharmacokinetics in Cynomolgus Monkeys
The in vivo pharmacokinetic properties of Example 1 are demonstrated using
cynomolgus monkeys. The compounds is administered by a single oral (PO; 10
mg/kg;
volume of 5 mL/kg) dose in vehicle. Blood is collected from each animal at 0,
0.25, 0.5, 1,
2, 4, 8, 12, 24, 32, 48, and 72 h post-dosage. The plasma concentrations of
Example 1 are
determined by a LC-MS/MS method as described above.
Example 1 mean half-life is 15.3 hours. This data shows Example 1 is orally
bioavailable and slowly eliminated in monkeys.
Intrinsic Clearance in Human Hepatocytes (-/+ ABT)
This method is intended to identify in vitro metabolic clearance by substrate
depletion
in hepatocytes. Incubation in the presence and absence of ABT, a pan-CYP450
enzyme
inhibitor, is used to estimate the contribution of CYP-mediated metabolism.
Cryopreserved
human hepatocytes are thawed at 37 C, spun down and reconstituted in
hepatocyte
maintenance media to a density of 1 x 106 viable cells/mL. To a pre-warmed 96-
well plate,
196 of the hepatocyte suspension is added to each well. The cells are
pre-incubated with
and without ABT as follows: for ABT pre-incubation, 2 tL of 100 mM ABT
solution is
added (2 tL of media without ABT is added to control samples), and the plate
incubated at
37 C for 30 min under constant shaking (-600 rpm). Following, 2 tL of a 30
i.tM stock
solution of test article is added and at 0, 15, 30, 60, 120, 240 min 20
aliquots are taken
and quenched by transfer to ACN containing internal standard. Following
centrifugation at
4000 rpm for 30 min, supernatant concentrations are determined by LC-MS/MS.
Clearance is
calculated from the slope of % compound remaining over time.

CA 03111725 2021-03-03
WO 2020/051058 PCT/US2019/048788
-25-
Clearance of Example 1 is inhibited by ABT by ¨12% in human hepatocytes,
suggesting limited involvement of CYP enzymes in the hepatic clearance of
Example 1.
Inhibition of OATP1B1 and OATP1B3
The OATP1B1, OATP1B3, and vector control (VC) cells are grown in 5% CO2 at 37
C in a humidified atmosphere in DMEM supplemented with 10% FBS, 50 [tg/mL
gentamycin, and 5 [tg/mL blasticidin. The cells are seeded in 24-well BioCoat
Poly-D-
Lysine plates. The cells are treated with 5 mM sodium butyrate in supplemented
DMEM 24
h prior to experimentation. Cell cultures are washed twice with pre-warmed PBS
prior to
experimentation. After washing, all cell types are incubated for 30 min at 37
C in 200 L
buffer, buffer plus varying concentration of test article, or with appropriate
positive control
inhibitor. Based upon preliminary data concentrations from 0.025 to 12.5 [tM
for OATP1B1
and 0.20 to 100 [tM for OATP1B3 (nominal) are tested. At the start of the
preincubation, 50
1..t.L of buffer is removed for determination of in-well test article
concentrations by LC-
MS/MS. Following the pre-incubation period, the buffer is removed. Experiments
are
initiated by the addition of 200 L substrate solution (400 nM rosuvastatin
total, including
1.4 nM tritiated rosuvastatin). Incubations are performed with and without
inhibitor.
Positive control inhibitor used for each cell line is 50 [tM rifamycin SV.
Experiments are
carried out for 1 min at 37 C, at which the reaction is stopped and washed
with the addition
of 1000 1..t.L ice cold PBS per well. Each well is then aspirated and washed
two additional
times with ice cold PBS. Cells in each well are lysed in 400 1..t.L 1% Triton
X 100 in PBS (by
volume). Samples are taken from each well from plates to count for
radioactivity and protein
concentration in each well is determined by bicinchoninic acid method. IC50
values are
determined by fitting the data using GraphPad Prism. Nominal concentrations
are converted
to measured concentrations prior to fitting.
Example 1 inhibited OATP1B1 more potently than OATP1B3, with IC50 values of
0.12 and 5.5 [tM, respectively.

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Letter Sent 2022-12-13
Grant by Issuance 2022-12-13
Inactive: Grant downloaded 2022-12-13
Inactive: Grant downloaded 2022-12-13
Inactive: Cover page published 2022-12-12
Inactive: Final fee received 2022-09-23
Pre-grant 2022-09-23
Notice of Allowance is Issued 2022-06-13
Letter Sent 2022-06-13
Notice of Allowance is Issued 2022-06-13
Inactive: Approved for allowance (AFA) 2022-05-05
Inactive: Q2 passed 2022-05-05
Common Representative Appointed 2021-11-13
Amendment Received - Voluntary Amendment 2021-04-28
Amendment Received - Voluntary Amendment 2021-04-28
Inactive: Cover page published 2021-03-25
Inactive: IPC removed 2021-03-22
Inactive: IPC assigned 2021-03-22
Inactive: IPC assigned 2021-03-22
Inactive: IPC assigned 2021-03-22
Inactive: IPC assigned 2021-03-22
Inactive: First IPC assigned 2021-03-22
Application Received - PCT 2021-03-17
Letter Sent 2021-03-17
Letter sent 2021-03-17
Priority Claim Requirements Determined Compliant 2021-03-17
Request for Priority Received 2021-03-17
Inactive: IPC assigned 2021-03-17
Inactive: IPC assigned 2021-03-17
Inactive: IPC assigned 2021-03-17
Inactive: IPC assigned 2021-03-17
Inactive: First IPC assigned 2021-03-17
National Entry Requirements Determined Compliant 2021-03-03
Request for Examination Requirements Determined Compliant 2021-03-03
All Requirements for Examination Determined Compliant 2021-03-03
Application Published (Open to Public Inspection) 2020-03-12

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2022-07-21

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2021-03-03 2021-03-03
Request for examination - standard 2024-08-29 2021-03-03
MF (application, 2nd anniv.) - standard 02 2021-08-30 2021-07-21
MF (application, 3rd anniv.) - standard 03 2022-08-29 2022-07-21
Final fee - standard 2022-10-13 2022-09-23
MF (patent, 4th anniv.) - standard 2023-08-29 2023-07-21
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
ELI LILLY AND COMPANY
Past Owners on Record
TIMOTHY BARRETT DURHAM
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2021-03-02 25 1,013
Claims 2021-03-02 3 69
Abstract 2021-03-02 1 52
Claims 2021-04-27 4 97
Courtesy - Letter Acknowledging PCT National Phase Entry 2021-03-16 1 593
Courtesy - Acknowledgement of Request for Examination 2021-03-16 1 435
Commissioner's Notice - Application Found Allowable 2022-06-12 1 576
Electronic Grant Certificate 2022-12-12 1 2,527
National entry request 2021-03-02 7 180
International search report 2021-03-02 2 60
Declaration 2021-03-02 2 31
Amendment / response to report 2021-04-27 8 182
Final fee 2022-09-22 3 66